1
|
Luo X, Luo L, Lai R, Li Y, Zhou H, Li X. Synthesis and Antioxidant Effects of Edaravone-Loaded MPEG-2000-DSPE Micelles in Rotenone-Induced PC12 Cell Model of Parkinson's Disease. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1962. [PMID: 39683350 DOI: 10.3390/nano14231962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder globally that lacks any disease-modifying drug for prevention or treatment. Oxidative stress has been identified as one of the key pathogenic drivers of Parkinson's disease (PD). Edaravone, an approved free-radical scavenger, has proven to have potential against PD by targeting multiple key pathologies, including oxidative stress, focal mitochondria, and neuroinflammation. However, its bioavailability is potentially restricted due to its poor solubility and short half-life. This study aims to develop a simple and effective drug delivery system for edaravone to enhance its solubility, stability, and bioavailability to improve its neuroprotective efficacy. An MPEG-2000-DSPE-edaravone (MDE) micelle was prepared via solvent evaporation using MPEG-2000-DSPE as a carrier to encapsulate edaravone. The morphology, particle size, zeta potential, chemical structure, and edaravone loading of MDE were evaluated. We then investigated whether such targeted edaravone delivery could provide enhanced neuroprotection. A cell model of PD was established in PC12 cells through exposure to rotenone. The effects of MDE on PC12 cells treated with or without rotenone were evaluated using a cell counting kit-8, calcein acetoxymethyl ester (AM)-propidine iodide (PI) staining, and flow cytometry. Cell migration was evaluated using a wound healing assay. Additionally, the intracellular antioxidant study was performed using an ROS-level-detecting DCFH-DA probe, and the mitochondrial membrane potentials were evaluated using a JC-1 assay. MDE with a drug-loading content of 17.6% and an encapsulation efficiency of 92.8% was successfully prepared. The resultant MDE had a mean particle size of 112.97 ± 5.54 nm with a zeta potential of -42 mV. Cytotoxicity assays confirmed that the MDE (≤200 ug/mL) exhibited promising cytocompatibility with no significant effect on cell viability, cell cycle regulation, or apoptosis levels. Likewise, compared with the free edaravone, no effect on cell migration was noted for MDE. MDE might be able to target edaravone delivery into PC12 cells, increasing the mitochondrial membrane potential and providing a significant local antioxidant effect. The results demonstrated that MPEG-2000-DSPE could be a promising material for enhancing edaravone's aqueous solubility, stability, and antioxidant effects. MDE could be a potential drug formulation for treating PD and other diseases in which oxidative stress plays a key role in pathogenesis.
Collapse
Affiliation(s)
- Xin Luo
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Linshan Luo
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Rong Lai
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Yan Li
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Hongyan Zhou
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xiting Li
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| |
Collapse
|
2
|
Patel D, Wairkar S. Effect of food on oral pharmacokinetics of edaravone coamorphous dispersion containing bile salts as coformers - Part II. ANNALES PHARMACEUTIQUES FRANÇAISES 2024; 82:1062-1070. [PMID: 38942078 DOI: 10.1016/j.pharma.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 06/30/2024]
Abstract
OBJECTIVES Edaravone (EDR) is an effective neuroprotective agent in various neurological diseases; however, its use is restricted due to poor oral absorption. Bile salts are known for improving solubility and inhibiting drug crystallization in supersaturated conditions of the gastrointestinal tract (GIT). In our previous work, we prepared coamorphous dispersion (COAM) of EDR with sodium taurocholate (NaTC) using spray drying. The optimized EDR COAM exhibited superior in vitro performance compared to plain EDR. EDR is well absorbed in fasted-over-fed conditions. METHODS The present work, we conducted a pharmacokinetic study for EDR and EDR COAM in fasted and fed conditions to check effect of food on its oral absorption. The LC-MS/MS-based method was developed and validated to determine the amount of EDR in plasma. RESULTS The results suggested that EDR COAM did not show a significant difference in Cmax (P=0.3544) and AUC (P=0.1696) of fasted and fed states. On the other hand, plain EDR showed 2-fold and 3-fold reduced Cmax (P<0.0001) and AUC (P=0.0094) in the fed condition, respectively. The Cmax and AUC of EDR COAM were improved in fasted (AUC: 2.56-fold) and fed states (AUC: 5.74-fold) than plain EDR, suggesting better oral absorption of COAM than crystalline EDR without having the effect of food. CONCLUSIONS The unique structural attributes of NaTC had the potential to inhibit the recrystallization of EDR in GIT, while concurrently reducing the impact of food on the oral absorption of EDR.
Collapse
Affiliation(s)
- Dhrumi Patel
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), 400056 Mumbai, Maharashtra, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L. Mehta Road, Vile Parle (W), 400056 Mumbai, Maharashtra, India.
| |
Collapse
|
3
|
O’Neill R, Yoo O, Burcham P, Lim LY. Edaravone for the Treatment of Motor Neurone Disease: A Critical Review of Approved and Alternative Formulations against a Proposed Quality Target Product Profile. Pharmaceutics 2024; 16:993. [PMID: 39204338 PMCID: PMC11360395 DOI: 10.3390/pharmaceutics16080993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/19/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Edaravone is one of two main drugs for treating motor neurone disease (MND). This review proposes a specific quality target product profile (QTPP) for edaravone following an appraisal of the issues accounting for the poor clinical uptake of the approved IV and oral liquid edaravone formulations. This is followed by a review of the alternative oral formulations of edaravone described in the published patent and journal literature against the QTPP. A total of 14 texts published by six research groups on 18 novel oral formulations of edaravone for the treatment of MND have been reviewed. The alternative oral formulations included liquid and solid formulations developed with cyclodextrins, lipids, surfactants, co-surfactants, alkalising agents, tablet excipients, and co-solvents. Most were intended to deliver edaravone for drug absorption in the lower gastrointestinal tract (GIT); however, there were also four formulations targeting the oral mucosal absorption of edaravone to avoid first-pass metabolism. All the novel formulations improved the aqueous solubility, stability, and oral bioavailability (BA) of edaravone compared to an aqueous suspension of edaravone. A common limitation of the published formulations is the lack of MND-patient-centred data. Except for TW001, no other formulations have been trialled in MND patients. To meet the QTPP of an oral edaravone formulation for MND patients, it is recommended that a tablet of appropriate size and with acceptable taste and stability be designed for the effective sublingual or buccal absorption of edaravone. This tablet should be designed with input from the MND community.
Collapse
Affiliation(s)
- Riuna O’Neill
- Division of Pharmacy, School of Allied Health, University of Western Australia, Perth, WA 6009, Australia; (R.O.); (O.Y.); (P.B.)
| | - Okhee Yoo
- Division of Pharmacy, School of Allied Health, University of Western Australia, Perth, WA 6009, Australia; (R.O.); (O.Y.); (P.B.)
- Institute for Paediatric Perioperative Excellence, University of Western Australia, Perth, WA 6009, Australia
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, WA 6009, Australia
| | - Philip Burcham
- Division of Pharmacy, School of Allied Health, University of Western Australia, Perth, WA 6009, Australia; (R.O.); (O.Y.); (P.B.)
- Division of Pharmacology and Toxicology, School of Biomedical Science, University of Western Australia, Perth, WA 6009, Australia
| | - Lee Yong Lim
- Division of Pharmacy, School of Allied Health, University of Western Australia, Perth, WA 6009, Australia; (R.O.); (O.Y.); (P.B.)
- Institute for Paediatric Perioperative Excellence, University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
4
|
Sanghai N, Vuong B, Burak Berk A, Afridi MSK, Tranmer GK. Current Small Molecule-Based Medicinal Chemistry Approaches for Neurodegeneration Therapeutics. ChemMedChem 2024; 19:e202300705. [PMID: 38329887 DOI: 10.1002/cmdc.202300705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/07/2024] [Accepted: 02/07/2024] [Indexed: 02/10/2024]
Abstract
Neurodegenerative diseases (NDDs) like Alzheimer's disease (AD), Parkinson's disease (PD), and Amyotrophic lateral sclerosis (ALS) possess multifactorial aetiologies. In recent years, our understanding of the biochemical and molecular pathways across NDDs has increased, however, new advances in small molecule-based therapeutic strategies targeting NDDs are obscure and scarce. Moreover, NDDs have been studied for more than five decades, however, there is a paucity of drugs that can treat NDDs. Further, the highly lipoidal blood-brain barrier (BBB) limits the uptake of many therapeutic molecules into the brain and is a complicating factor in the development of new agents to treat neurodegeneration. Considering the highly complex nature of NDDs, the association of multiple risk factors, and the challenges to overcome the BBB junction, medicinal chemists have developed small organic molecule-based novel approaches to target NDDs over the last few decades, such as designing lipophilic molecules and applying prodrug strategies. Attempts have been made to utilize a multitarget approach to modulate different biochemical molecular pathways involved in NDDs, in addition to, medicinal chemists making better decisions in identifying optimized drug candidates for the central nervous system (CNS) by using web-based computational tools. To increase the clinical success of these drug candidates, an in vitro assay modeling the BBB has been utilized by medicinal chemists in the pre-clinical phase as a further screening measure of small organic molecules. Herein, we examine some of the intriguing strategies taken by medicinal chemists to design small organic molecules to combat NDDs, with the intention of increasing our awareness of neurodegenerative therapeutics.
Collapse
Affiliation(s)
- Nitesh Sanghai
- College of Pharmacy, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Billy Vuong
- College of Pharmacy, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | - Ahmet Burak Berk
- College of Pharmacy, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| | | | - Geoffrey K Tranmer
- College of Pharmacy, Rady Faculty of Health Science, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| |
Collapse
|
5
|
Patel D, Wairkar S. In silico and in vitro investigation of bile salts as coformers for edaravone coamorphous dispersion- Part I. Chem Phys Lipids 2023; 253:105302. [PMID: 37031754 DOI: 10.1016/j.chemphyslip.2023.105302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 04/11/2023]
Abstract
In the present study, we aimed to design the spray-dried coamorphous dispersion (COAM) of a neuroprotective agent-edaravone (EDR) with bile salts to improve oral bioavailability. After the initial screening of different bile salts, EDR-sodium taurocholate (NaTC) COAM showed 4-fold solubility than a pure drug in 1-7pH range. In silico studies to select coformer for COAM revealed a narrow energy gap, easy charge transfer and high chemical reactivity between EDR and NaTC. The optimized EDR-NaTC COAM in a 1:1 molar ratio was characterized for solid state characterizations and in vitro release study. Hydrogen bond formation between the pyrazolone ring of EDR and the -OH group of the phenanthrene ring of NaTC was observed in the ATR-FTIR spectra of COAM. The DSC and XRPD data indicated the formation of an amorphous halo, whereas SEM photographs demonstrated porous, spherical particles of COAM. The pH-independent in vitro drug release of COAM was observed in 0.1N HCl, pH 4.5 and 6.8 buffers which was 3-fold higher than EDR. The COAM was stable for 6 months at accelerated condition without showing a change in drug content or devitrification (Initial: 98.002±0.942%; Accelerated condition: 97.016±1.110%). Although coamorphous form and hydrogen bonding between EDR-NaTC dispersion were primarily responsible for improved dissolution, NaTC, an exceptional surfactant, has also contributed to it. Moreover, its exclusive structural characteristics could prevent the recrystallization of the drug in supersaturated conditions of the GIT and also minimize the effect of food on oral absorption of EDR which will be studied in animals in the second part of this work.
Collapse
Affiliation(s)
- Dhrumi Patel
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai. Maharashtra - 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai. Maharashtra - 400056, India.
| |
Collapse
|
6
|
Liu Z, Lansley AB, Duong TN, Smart JD, Pannala AS. Increasing Cellular Uptake and Permeation of Curcumin Using a Novel Polymer-Surfactant Formulation. Biomolecules 2022; 12:biom12121739. [PMID: 36551167 PMCID: PMC9775279 DOI: 10.3390/biom12121739] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/16/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Several therapeutically active molecules are poorly water-soluble, thereby creating a challenge for pharmaceutical scientists to develop an active solution for their oral drug delivery. This study aimed to investigate the potential for novel polymer-surfactant-based formulations (designated A and B) to improve the solubility and permeability of curcumin. A solubility study and characterization studies (FTIR, DSC and XRD) were conducted for the various formulations. The cytotoxicity of formulations and commercial comparators was tested via MTT and LDH assays, and their permeability by in vitro drug transport and cellular drug uptake was established using the Caco-2 cell model. The apparent permeability coefficients (Papp) are considered a good indicator of drug permeation. However, it can be argued that the magnitude of Papp, when used to reflect the permeability of the cells to the drug, can be influenced by the initial drug concentration (C0) in the donor chamber. Therefore, Papp (suspension) and Papp (solution) were calculated based on the different values of C0. It was clear that Papp (solution) can more accurately reflect drug permeation than Papp (suspension). Formulation A, containing Soluplus® and vitamin E TPGs, significantly increased the permeation and cellular uptake of curcumin compared to other samples, which is believed to be related to the increased aqueous solubility of the drug in this formulation.
Collapse
Affiliation(s)
- Zhenqi Liu
- Biomaterials and Drug Delivery Research Group, School of Applied Sciences, University of Brighton, Brighton BN2 4GJ, UK
| | - Alison B. Lansley
- Biomaterials and Drug Delivery Research Group, School of Applied Sciences, University of Brighton, Brighton BN2 4GJ, UK
| | - Tu Ngoc Duong
- Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, Hanoi 100000, Vietnam
| | - John D. Smart
- Biomaterials and Drug Delivery Research Group, School of Applied Sciences, University of Brighton, Brighton BN2 4GJ, UK
| | - Ananth S. Pannala
- Biomaterials and Drug Delivery Research Group, School of Applied Sciences, University of Brighton, Brighton BN2 4GJ, UK
- Correspondence:
| |
Collapse
|
7
|
Du J, Hong Y, Cheng L, Gu Z, Li Z, Li C. Effects of acid-ethanol hydrolysis and debranch on acetylated starch and its potential used for curcumin carrier. Carbohydr Polym 2022; 279:119019. [PMID: 34980359 DOI: 10.1016/j.carbpol.2021.119019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/12/2021] [Indexed: 11/28/2022]
Abstract
Acetylated acid-ethanol hydrolyzed (AHS) and acetylated debranched starch (ADS) were investigated as prospective nanocarriers. Both acid-ethanol hydrolysis and debranching decreased the molecular weight and viscosity of starch. Acid-ethanol hydrolyzed starch remained the original microstructure, which was confirmed by results of scanning electron microscopy. New absorption peaks in FTIR spectra of starch confirmed the occurrence of acetylation. The substitution degree (DS) of ADS could reach up to 1.18, while that of AHS could be improved by increasing the ethanol concentration. The developed nanoparticles showed uniform spherical structure and the size of that approximated 180-260 nm. The critical micelle concentration was 0.049 mg/mL, and the shift in fluorescence spectra confirmed the interaction between starch and curcumin. These results indicate show that high DS of AHS and ADS could be used as a potential carrier for curcumin delivery.
Collapse
Affiliation(s)
- Jing Du
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi 214122, Jiangsu Province, People's Republic of China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Yan Hong
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi 214122, Jiangsu Province, People's Republic of China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China.
| | - Li Cheng
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi 214122, Jiangsu Province, People's Republic of China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Zhengbiao Gu
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi 214122, Jiangsu Province, People's Republic of China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Zhaofeng Li
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi 214122, Jiangsu Province, People's Republic of China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| | - Caiming Li
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Wuxi 214122, Jiangsu Province, People's Republic of China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China; Collaborative Innovation Center for Food Safety and Quality Control, Jiangnan University, Wuxi 214122, Jiangsu Province, People's Republic of China
| |
Collapse
|
8
|
Avdeef A, Kansy M. Predicting Solubility of Newly-Approved Drugs (2016–2020) with a Simple ABSOLV and GSE(Flexible-Acceptor) Consensus Model Outperforming Random Forest Regression. J SOLUTION CHEM 2022; 51:1020-1055. [PMID: 35153342 PMCID: PMC8818506 DOI: 10.1007/s10953-022-01141-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/10/2021] [Indexed: 11/24/2022]
Abstract
This study applies the ‘Flexible-Acceptor’ variant of the General Solubility Equation, GSE(Φ,B), to the prediction of the aqueous intrinsic solubility, log10S0, of FDA recently-approved (2016–2020) ‘small-molecule’ new molecular entities (NMEs). The novel equation had been shown to predict the solubility of drugs beyond Lipinski’s ‘Rule of 5’ chemical space (bRo5) to a precision nearly matching that of the Random Forest Regression (RFR) machine learning method. Since then, it was found that the GSE(Φ,B) appears to work well not only for bRo5 NMEs, but also for Ro5 drugs. To put context to GSE(Φ,B), Yalkowsky’s GSE(classic), Abraham’s ABSOLV, and Breiman’s RFR models were also applied to predict log10 S0 of 72 newly-approve NMEs, for which useable reported solubility values could be accessed (nearly 60% from FDA New Drug Application published reports). Except for GSE (classic), the prediction models were retrained with an enlarged version of the Wiki-pS0 database (nearly 400 added log10 S0 entries since our recent previous study). Thus, these four models were further validated by the additional independent solubility measurements which the newly-approved drugs introduced. The prediction methods ranked RFR ~ GSE (Φ,B) > ABSOLV > GSE (classic) in performance. It was further demonstrated that the biases generated in the four separate models could be nearly eliminated in a consensus model based on the average of just two of the methods: GSE (Φ,B) and ABSOLV. The resulting consensus prediction equation is simple in form and can be easily incorporated into spreadsheet calculations. Even more significant, it slightly outperformed the RFR method.
Collapse
|
9
|
Solubility and Dissolution Enhancement of Dexibuprofen with Hydroxypropylbetacyclodextrin (HPβCD) and Poloxamers (188/407) Inclusion Complexes: Preparation and In Vitro Characterization. Polymers (Basel) 2022; 14:polym14030579. [PMID: 35160569 PMCID: PMC8838044 DOI: 10.3390/polym14030579] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 02/02/2023] Open
Abstract
The objective of this study was to improve the dissolution and solubility of dexibuprofen (DEX) using hydroxypropyl beta cyclodextrin (HPβCD) inclusion complexes and also to evaluate the effect of presence of hydrophilic polymers on solubilization efficiency of HPβCD. Three different methods (physical trituration, kneading and solvent evaporation) were used to prepare binary inclusion complexes at various drug-to-cyclodextrin weight ratios. An increase in solubility and drug release was observed with the kneading (KN) method at a DEX/HPβCD (1:4) weight ratio. The addition of hydrophilic polymers poloxamer-188 (PXM-188) and poloxamer-407 (PXM-407) at 2.5, 5.0, 10.0 and 20% w/w enhanced the complexation efficiency and solubility of DEX/HPβCD significantly. Fourier-transform infrared (FTIR) analysis revealed that DEX was successfully incorporated into the cyclodextrin cavity. Differential scanning calorimetry (DSC) and X-ray diffractometry (XRD) revealed less crystallinity of the drug and its entrapment in the cyclodextrin molecular cage. The addition of PXM-188 or PXM-407 reduced the strength of the DEX endothermic peak. With the addition of hydrophilic polymers, sharp and intense peaks of DEX disappeared. Finally, it was concluded that PXM-188 at a weight ratio of 10.0% w/w was the best candidate for improving solubility, stability and release rate of DEX.
Collapse
|
10
|
Hiroshige R, Goto S, Ichii R, Shimizu S, Wada-Hirai A, Li YP, Shimada Y, Otsuka Y, Makino K, Takahashi H. Protective effects of cyclodextrins on edaravone degradation induced by atmospheric oxygen or additive oxidant. J INCL PHENOM MACRO 2021. [DOI: 10.1007/s10847-021-01122-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
11
|
Preclinical validation of a novel oral Edaravone formulation for treatment of frontotemporal dementia. Neurotox Res 2021; 39:1689-1707. [PMID: 34599751 DOI: 10.1007/s12640-021-00405-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 10/20/2022]
Abstract
Oxidative stress is a key factor in the pathogenesis of several neurodegenerative disorders and is involved in the accumulation of amyloid beta plaques and Tau inclusions. Edaravone (EDR) is a free radical scavenger that is approved for motor neuron disease and acute ischemic stroke. EDR alleviates pathologies and cognitive impairment of AD via targeting multiple key pathways in transgenic mice. Herein, we aimed to study the effect of EDR on Tau pathology in P301L mice; an animal model of frontotemporal dementia (FTD), at two age time points representing the early and late stages of the disease. A novel EDR formulation was utilized in the study and the drug was delivered orally in drinking water for 3 months. Then, behavioral tests were conducted followed by animal sacrifice and brain dissection. Treatment with EDR improved the reference memory and accuracy in the probe trial as evaluated in Morris water maze, as well as novel object recognition and significantly alleviated motor deficits in these mice. EDR also reduced the levels of 4-hydroxy-2-nonenal and 3-nitrotyrosine adducts. In addition, immunohistochemistry showed that EDR reduced tau phosphorylation and neuroinflammation and partially rescued neurons against oxidative neurotoxicity. Moreover, EDR attenuated downstream pathologies involved in Tau hyperphosphorylation. These results suggest that EDR may be a potential therapeutic agent for the treatment of FTD.
Collapse
|
12
|
Wang W, Ouyang D. Prediction of Free Drug Absorption in Cyclodextrin Formulation by a Modified Physiologically Based Pharmacokinetic Model and Phase Solubility 3-D Surface Graph. Pharm Res 2021; 38:1157-1168. [PMID: 34145531 DOI: 10.1007/s11095-021-03071-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 06/01/2021] [Indexed: 12/20/2022]
Abstract
PURPOSE Cyclodextrin (CD) is commonly used to enhance the solubility of oral drugs. However, with the increase of CD concentrations, the fraction of free drug molecules decreases, which may potentially impede drug absorption. This study aims to predict the optimal ratio between drug and CD to achieve the best absorption efficiency by computational simulation. METHODS First, a physiologically based pharmacokinetic (PBPK) model was developed. This model can continuously adjust absorption according to free drug fraction and was validated against two model drugs, progesterone (PG) and andrographolide (AG). The further analysis involves 3-D surface graphs to investigate the relationship between free drug amount, theoretically absorbable concentration, and contents of drug and CD in the formulation. RESULTS The PBPK model predicted the PK behavior of two drugs well. The concentration ratio of drug to CD, leading to maximal free drug amount and the best absorption efficiency, is nearly the same as the slope determined in the phase solubility test. The new modified PBPK model and 3-D surface graph can easily predict the absorption difference of formulations with various drug/CD ratios. CONCLUSION This PBPK model and 3-D surface graph can predict the absorption and determine the optimal concentration ratio of CD formulation, which could accelerate the R&D of CD formulation.
Collapse
Affiliation(s)
- Wei Wang
- Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Defang Ouyang
- Institute of Chinese Medical Sciences, University of Macau, Macau, China.
| |
Collapse
|
13
|
Enhanced solubility, stability, permeation and anti-cancer efficacy of Celastrol-β-cyclodextrin inclusion complex. J Mol Liq 2020. [DOI: 10.1016/j.molliq.2020.113936] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
14
|
Hydroxy-Propil-β-Cyclodextrin Inclusion Complexes of two Biphenylnicotinamide Derivatives: Formulation and Anti-Proliferative Activity Evaluation in Pancreatic Cancer Cell Models. Int J Mol Sci 2020; 21:ijms21186545. [PMID: 32906812 PMCID: PMC7576480 DOI: 10.3390/ijms21186545] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 01/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies, with poor outcomes largely due to its unique microenvironment, which is responsible for the low response to drugs and drug-resistance phenomena. This clinical need led us to explore new therapeutic approaches for systemic PDAC treatment by the utilization of two newly synthesized biphenylnicotinamide derivatives, PTA73 and PTA34, with remarkable antitumor activity in an in vitro PDAC model. Given their poor water solubility, inclusion complexes of PTA34 and PTA73 in Hydroxy-Propil-β-Cyclodextrin (HP-β-CD) were prepared in solution and at the solid state. Complexation studies demonstrated that HP-β-CD is able to form stable host–guest inclusion complexes with PTA34 and PTA73, characterized by a 1:1 apparent formation constant of 503.9 M−1 and 369.2 M−1, respectively (also demonstrated by the Job plot), and by an increase in aqueous solubility of about 150 times (from 1.95 µg/mL to 292.5 µg/mL) and 106 times (from 7.16 µg/mL to 762.5 µg/mL), in the presence of 45% w/v of HP-β-CD, respectively. In vitro studies confirmed the high antitumor activity of the complexed PTA34 and PTA73 towards PDAC cells, the strong G2/M phase arrest followed by induction of apoptosis, and thus their eligibility for PDAC therapy.
Collapse
|
15
|
Bashir M, Syed HK, Asghar S, Irfan M, Almalki WH, Menshawi SA, Khan IU, Shah PA, Khalid I, Ahmad J, Gohar UF, Peh KK, Iqbal MS. Effect of Hydrophilic Polymers on Complexation Efficiency of Cyclodextrins in Enhancing Solubility and Release of Diflunisal. Polymers (Basel) 2020; 12:E1564. [PMID: 32679660 PMCID: PMC7408593 DOI: 10.3390/polym12071564] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/04/2020] [Accepted: 07/07/2020] [Indexed: 02/05/2023] Open
Abstract
The effects of three hydrophilic polymers, namely, carboxymethyl cellulose sodium (CMC-Na), polyvinyl alcohol (PVA) and poloxamer-188 (PXM-188) on the solubility and dissolution of diflunisal (DIF) in complexation with β-cyclodextrin (βCD) or hydroxypropyl β-cyclodextrin (HPβCD), were investigated. The kneading method was used at different drug to cyclodextrin weight ratios. Increases in solubility and drug release were observed with the DIF/βCD and DIF/HPβCD complexes. The addition of hydrophilic polymers at 2.5, 5.0 and 10.0% w/w markedly improved the complexation and solubilizing efficiency of βCD and HPβCD. Fourier-transform infrared (FTIR) showed that DIF was successfully included into the cyclodextrin cavity. Differential scanning calorimetry (DSC) and X-ray diffractometry (XRD) confirmed stronger drug amorphization and entrapment in the molecular cage of cyclodextrins. The addition of PVA, CMC-Na or PXM-188 reduced further the intensity of the DIF endothermic peak. Most of the sharp and intense peaks of DIF disappeared with the addition of hydrophilic polymers. In conclusion, PXM-188 at a weight ratio of 10.0% w/w was the best candidate in enhancing the solubility, stability and release of DIF.
Collapse
Affiliation(s)
- Mehreen Bashir
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan; (M.B.); (S.A.); (M.I.); (I.U.K.); (I.K.); (J.A.)
| | - Haroon Khalid Syed
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan; (M.B.); (S.A.); (M.I.); (I.U.K.); (I.K.); (J.A.)
| | - Sajid Asghar
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan; (M.B.); (S.A.); (M.I.); (I.U.K.); (I.K.); (J.A.)
| | - Muhammad Irfan
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan; (M.B.); (S.A.); (M.I.); (I.U.K.); (I.K.); (J.A.)
| | - Waleed Hassan Almalki
- Department of Toxicology and Pharmacology, College of Pharmacy, Umm Al Qura University, Makkah 21955, Saudi Arabia;
| | - Salah Ali Menshawi
- Department of Toxicology in Comprehensive Specialized Clinics Security Forces, Jeddah 21442, Saudi Arabia;
| | - Ikram Ullah Khan
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan; (M.B.); (S.A.); (M.I.); (I.U.K.); (I.K.); (J.A.)
| | - Pervaiz A. Shah
- University College of Pharmacy, University of the Punjab, Lahore 54590, Pakistan;
| | - Ikrima Khalid
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan; (M.B.); (S.A.); (M.I.); (I.U.K.); (I.K.); (J.A.)
| | - Junaid Ahmad
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan; (M.B.); (S.A.); (M.I.); (I.U.K.); (I.K.); (J.A.)
| | - Umar Farooq Gohar
- Institute of Industrial Biotechnology, Government College University, Lahore 54590, Pakistan;
| | - Kok Khiang Peh
- Department of Pharmaceutical Technology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia
| | - Muhammad Shahid Iqbal
- Department of Clinical Pharmacy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia;
| |
Collapse
|
16
|
Tang B, Qian Y, Fang G. Development of Lipid-Polymer Hybrid Nanoparticles for Improving Oral Absorption of Enoxaparin. Pharmaceutics 2020; 12:E607. [PMID: 32629827 PMCID: PMC7407632 DOI: 10.3390/pharmaceutics12070607] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/18/2020] [Accepted: 06/25/2020] [Indexed: 01/31/2023] Open
Abstract
Enoxaparin, an anticoagulant that helps prevent the formation of blood clots, is administered parenterally. Here, we report the development and evaluation of lipid-polymer hybrid nanoparticles (LPHNs) for the oral delivery of enoxaparin. The polymer poloxamer 407 (P407) was incorporated into lipid nanoparticles to form gel cores and ensure high encapsulation efficiency and the controlled release of enoxaparin. In vitro results indicated that 30% of P407 incorporation offered higher encapsulation efficiency and sustained the release of enoxaparin. Laser confocal scanning microscopy (LCSM) images showed that LPHNs could not only significantly improve the accumulation of enoxaparin in intestinal villi but also facilitate enoxaparin transport into the underlayer of intestinal epithelial cells. In vivo pharmacokinetic study results indicated that the oral bioavailability of enoxaparin was markedly increased about 6.8-fold by LPHNs. In addition, its therapeutic efficacy against pulmonary thromboembolism was improved 2.99-fold by LPHNs. Moreover, LPHNs exhibited excellent biocompatibility in the intestine. Overall, the LPHN is a promising delivery carrier to boost the oral absorption of enoxaparin.
Collapse
Affiliation(s)
- Bo Tang
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong 226001, China; (B.T.); (Y.Q.)
- School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
- Yabang Medical Research Institute, 66 Changhong Road, Changzhou 213145, China
| | - Yu Qian
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong 226001, China; (B.T.); (Y.Q.)
| | - Guihua Fang
- School of Pharmacy, Nantong University, 19 Qixiu Road, Nantong 226001, China; (B.T.); (Y.Q.)
| |
Collapse
|
17
|
Dash RP, Babu RJ, Srinivas NR. Two Decades-Long Journey from Riluzole to Edaravone: Revisiting the Clinical Pharmacokinetics of the Only Two Amyotrophic Lateral Sclerosis Therapeutics. Clin Pharmacokinet 2019; 57:1385-1398. [PMID: 29682695 DOI: 10.1007/s40262-018-0655-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The recent approval of edaravone has provided an intravenous option to treat amyotrophic lateral sclerosis (ALS) in addition to the existing oral agent, riluzole. The present work was primarily undertaken to provide a comprehensive clinical pharmacokinetic summary of the two approved ALS therapeutics. The key objectives of the review were to (i) tabulate the clinical pharmacokinetics of riluzole and edaravone with emphasis on absorption, distribution, metabolism and excretion (ADME) properties; (ii) provide a comparative scenario of the pharmacokinetics of the two drugs wherever possible; and (iii) provide perspectives and introspection on the gathered clinical pharmacokinetic data of the two drugs with appropriate conjectures to quench scientific curiosity. Based on this review, the following key highlights were deduced: (i) as a result of both presystemic metabolism and polymorphic hepatic cytochrome P450 (CYP) metabolism, the oral drug riluzole exhibited more inter-subject variability than that of intravenous edaravone; (ii) using various parameters for comparison, including the published intravenous data for riluzole, it was apparent that edaravone was achieving the desired systemic concentrations to possibly drive the local brain concentrations for its efficacy in ALS patients with lesser variability than riluzole; (iii) using scientific conjectures, it was deduced that the availability of intravenous riluzole may not be beneficial in therapy due to its fast systemic clearance; (iv) on the contrary, however, there appeared to be an opportunity for the development of an oral dosage form of edaravone, which may potentially benefit the therapy option for ALS patients by avoiding hospitalization costs; and (v) because of the existence of pharmaco-resistance for the brain entry in ALS patients, it appeared prudent to consider combination strategies of edaravone and/or riluzole with suitable P-glycoprotein efflux-blocking drugs to gain more favorable outcomes in ALS patients.
Collapse
Affiliation(s)
- Ranjeet Prasad Dash
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - R Jayachandra Babu
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Nuggehally R Srinivas
- Drug Metabolism and Pharmacokinetics, Zydus Research Centre, Ahmedabad, Gujarat, 382210, India.
| |
Collapse
|
18
|
Rat intestinal drug permeability: A status report and summary of repeated determinations. Eur J Pharm Biopharm 2019; 142:364-376. [DOI: 10.1016/j.ejpb.2019.07.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 06/28/2019] [Accepted: 07/04/2019] [Indexed: 12/28/2022]
|
19
|
Rashid M, Malik MY, Singh SK, Chaturvedi S, Gayen JR, Wahajuddin M. Bioavailability Enhancement of Poorly Soluble Drugs: The Holy Grail in Pharma Industry. Curr Pharm Des 2019; 25:987-1020. [DOI: 10.2174/1381612825666190130110653] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 01/24/2019] [Indexed: 02/07/2023]
Abstract
Background:
Bioavailability, one of the prime pharmacokinetic properties of a drug, is defined as the
fraction of an administered dose of unchanged drug that reaches the systemic circulation and is used to describe
the systemic availability of a drug. Bioavailability assessment is imperative in order to demonstrate whether the
drug attains the desirable systemic exposure for effective therapy. In recent years, bioavailability has become
the subject of importance in drug discovery and development studies.
Methods:
A systematic literature review in the field of bioavailability and the approaches towards its enhancement
have been comprehensively done, purely focusing upon recent papers. The data mining was performed
using databases like PubMed, Science Direct and general Google searches and the collected data was exhaustively
studied and summarized in a generalized manner.
Results:
The main prospect of this review was to generate a comprehensive one-stop summary of the numerous
available approaches and their pharmaceutical applications in improving the stability concerns, physicochemical
and mechanical properties of the poorly water-soluble drugs which directly or indirectly augment their bioavailability.
Conclusion:
The use of novel methods, including but not limited to, nano-based formulations, bio-enhancers,
solid dispersions, lipid-and polymer-based formulations which provide a wide range of applications not only
increases the solubility and permeability of the poorly bioavailable drugs but also improves their stability, and
targeting efficacy. Although, these methods have drastically changed the pharmaceutical industry demand for the
newer potential methods with better outcomes in the field of pharmaceutical science to formulate various dosage
forms with adequate systemic availability and improved patient compliance, further research is required.
Collapse
Affiliation(s)
- Mamunur Rashid
- Pharmaceutics and Pharmacokinetics Division, CSIR-CDRI, Lucknow, India
| | - Mohd Yaseen Malik
- Pharmaceutics and Pharmacokinetics Division, CSIR-CDRI, Lucknow, India
| | - Sandeep K. Singh
- Pharmaceutics and Pharmacokinetics Division, CSIR-CDRI, Lucknow, India
| | - Swati Chaturvedi
- Pharmaceutics and Pharmacokinetics Division, CSIR-CDRI, Lucknow, India
| | - Jiaur R Gayen
- Pharmaceutics and Pharmacokinetics Division, CSIR-CDRI, Lucknow, India
| | | |
Collapse
|
20
|
Cyclodextrin-membrane interaction in drug delivery and membrane structure maintenance. Int J Pharm 2019; 564:59-76. [DOI: 10.1016/j.ijpharm.2019.03.063] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 01/14/2023]
|
21
|
Kong R, Zhu X, Meteleva ES, Polyakov NE, Khvostov MV, Baev DS, Tolstikova TG, Dushkin AV, Su W. Atorvastatin calcium inclusion complexation with polysaccharide arabinogalactan and saponin disodium glycyrrhizate for increasing of solubility and bioavailability. Drug Deliv Transl Res 2018; 8:1200-1213. [PMID: 30039497 DOI: 10.1007/s13346-018-0565-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The aim of the present investigation was to enhance the solubility and dissolution of atorvastatin calcium (ATV), a poorly water-soluble drug with larch polysaccharide arabinogalactan (AG) and disodium glycyrrhizate (Na2GA) as carriers of drug delivery systems for improving its bioavailability. The interactions of ATV with AG or Na2GA were investigated by DSC, XRD, SEM, and NMR techniques. The molecular weights of supramolecular systems-inclusion complexes and micelles-which are the hosts for ATV molecules were measured. On the other hand, the rapid storage assay (+ 40 °C for 3 months) showed that the chemical stability of ATV/AG and ATV/Na2GA complexes had been enhanced compared with pure ATV. In vitro drug release showed a significant increase in ATV's dissolution rate after formation of a complex with Na2GA or AG. Pharmacokinetic tests in vivo on laboratory animals showed a significant increase in ATV's bioavailability after its introduction as a complex with Na2GA or AG. Moreover, ATV/AG and ATV/Na2GA complexes showed a more prominent decrease of total cholesterol (TC) level compared to net ATV. Therefore, the novel mechanochemically synthesized complexes of ATV with AG or Na2GA as drug delivery systems might be potential and promising candidates for hypercholesterolemia treatment and deserved further researches.
Collapse
Affiliation(s)
- Ruiping Kong
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Xingyi Zhu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Elizaveta S Meteleva
- Institute of Solid State Chemistry and Mechanochemistry, SB RAS, Novosibirsk, Russia, 630128
| | - Nikolay E Polyakov
- Institute of Chemical Kinetics and Combustion, SB RAS, Novosibirsk, Russia
| | - Mikhail V Khvostov
- N.N. Vorozhtsov Institute of Organic Chemistry, SB RAS, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Dmitry S Baev
- N.N. Vorozhtsov Institute of Organic Chemistry, SB RAS, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Tatjana G Tolstikova
- N.N. Vorozhtsov Institute of Organic Chemistry, SB RAS, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Alexander V Dushkin
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China.
- Institute of Solid State Chemistry and Mechanochemistry, SB RAS, Novosibirsk, Russia, 630128.
| | - Weike Su
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China.
| |
Collapse
|
22
|
Vaidya B, Shukla SK, Kolluru S, Huen M, Mulla N, Mehra N, Kanabar D, Palakurthi S, Ayehunie S, Muth A, Gupta V. Nintedanib-cyclodextrin complex to improve bio-activity and intestinal permeability. Carbohydr Polym 2018; 204:68-77. [PMID: 30366544 DOI: 10.1016/j.carbpol.2018.09.080] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 09/28/2018] [Accepted: 09/28/2018] [Indexed: 11/19/2022]
Abstract
Cyclodextrin complex of nintedanib was prepared aiming for increased bio-activity and improved transport across intestinal membrane with reduced p-glycoprotein (p-gp) efflux. Based on preliminary phase solubility studies and molecular modeling, sulfobutyl ether derivative of β-cyclodextrin (SBE-β-CD, Captisol®) was selected to prepare inclusion complex. Complexation was confirmed using FTIR, 1H NMR, DSC, and XRD. Bioactivity of the formed complex was tested using lung fibroblast cells, WI-38 for anti-proliferative activity and effect on collagen deposition and cells migration. In-vitro permeability studies were performed using epiIntestinal tissue model to assess the effect of complexation on transport and p-gp efflux. Results of the study demonstrated that cyclodextrin complexation increased stability of nintedanib in PBS (pH 7.4) and simulated intestinal fluid (SIF). Further, bioactivity of nintedanib also improved. Interestingly, complexation has increased transport of nintedanib across intestinal membrane and reduced efflux ratio, suggesting the role of cyclodextrin complexation in modulating p-gp efflux.
Collapse
Affiliation(s)
- Bhuvaneshwar Vaidya
- School of Pharmacy, Keck Graduate Institute, Claremont, CA 91711, United States
| | - Snehal K Shukla
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, United States
| | - Srikanth Kolluru
- School of Pharmacy, Keck Graduate Institute, Claremont, CA 91711, United States
| | - Melanie Huen
- School of Pharmacy, Keck Graduate Institute, Claremont, CA 91711, United States
| | - Nihal Mulla
- College of Pharmacy and Health Sciences, Drake University, Des Moines, IA 50311, United States
| | - Neelesh Mehra
- Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX 78363, United States
| | - Dipti Kanabar
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, United States
| | - Srinath Palakurthi
- Irma Lerma Rangel College of Pharmacy, Texas A&M Health Science Center, Kingsville, TX 78363, United States
| | | | - Aaron Muth
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, United States
| | - Vivek Gupta
- School of Pharmacy, Keck Graduate Institute, Claremont, CA 91711, United States; College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, United States.
| |
Collapse
|
23
|
Guimaraes PPG, Tan M, Tammela T, Wu K, Chung A, Oberli M, Wang K, Spektor R, Riley RS, Viana CTR, Jacks T, Langer R, Mitchell MJ. Potent in vivo lung cancer Wnt signaling inhibition via cyclodextrin-LGK974 inclusion complexes. J Control Release 2018; 290:75-87. [PMID: 30290244 DOI: 10.1016/j.jconrel.2018.09.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/30/2018] [Accepted: 09/25/2018] [Indexed: 01/02/2023]
Abstract
Activation of the Wnt signaling pathway promotes lung cancer progression and contributes to poor patient prognosis. The porcupine inhibitor LGK974, a novel orally bioavailable cancer therapeutic in Phase I clinical trials, induces potent Wnt signaling inhibition and leads to suppressed growth and progression of multiple types of cancers. The clinical use of LGK974, however, is limited in part due to its low solubility and high toxicity in tissues that rely on Wnt signaling for normal homeostasis. Here, we report the use of host-guest chemistry to enhance the solubility and bioavailability of LGK974 in mice through complexation with cyclodextrins (CD). We assessed the effects of these complexes to inhibit Wnt signaling in lung adenocarcinomas that are typically driven by overactive Wnt signaling. 2D 1H NMR confirmed host-guest complexation of CDs with LGK974. CD:LGK974 complexes significantly decreased the expression of Wnt target genes in lung cancer organoids and in lung cancer allografts in mice. Further, CD:LGK974 complexes increased the bioavailability upon oral administration in mice compared to free LGK974. In a mouse lung cancer allograft model, CD:LGK974 complexes induced potent Wnt signaling inhibition with reduced intestinal toxicity compared to treatment with free drug. Collectively, the development of these complexes enables safer and repeated oral or parenteral administration of Wnt signaling inhibitors, which hold promise for the treatment of multiple types of malignancies.
Collapse
Affiliation(s)
- Pedro P G Guimaraes
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, United States; Department of Chemical Engineering, MIT, Cambridge, MA, United States; Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Mingchee Tan
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, United States; Department of Chemical Engineering, MIT, Cambridge, MA, United States
| | - Tuomas Tammela
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, United States
| | - Katherine Wu
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, United States
| | - Amanda Chung
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, United States; Department of Chemical Engineering, MIT, Cambridge, MA, United States
| | - Matthias Oberli
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, United States; Department of Chemical Engineering, MIT, Cambridge, MA, United States
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Roman Spektor
- Graduate Field of Genetics, Genomics and Development, Cornell University, Ithaca, NY, United States
| | - Rachel S Riley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Celso T R Viana
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Tyler Jacks
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, United States
| | - Robert Langer
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, United States; Department of Chemical Engineering, MIT, Cambridge, MA, United States.
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
24
|
Identification of the primary determining factor(s) governing the oral absorption of edaravone in rats. Eur J Pharm Sci 2018; 123:312-320. [DOI: 10.1016/j.ejps.2018.07.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 07/04/2018] [Accepted: 07/26/2018] [Indexed: 12/19/2022]
|
25
|
Wang J, Chen X, Yuan B, Wang W, Xu C, Zhao W, Zhao P, Wang Y, Zhao X, Wang Y. Bioavailability of Edaravone Sublingual Tablet Versus Intravenous Infusion in Healthy Male Volunteers. Clin Ther 2018; 40:1683-1691. [PMID: 30241688 DOI: 10.1016/j.clinthera.2018.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/03/2018] [Accepted: 08/13/2018] [Indexed: 01/20/2023]
Abstract
PURPOSE Edaravone is a free-radical scavenger. Edaravone 30mg IV has been approved for use in the treatment of acute ischemic stroke in Japan and China, and for amyotrophic lateral sclerosis in Japan and the United States. Considering the inconvenience of IV infusion in clinical practice, an oral tablet formulation of edaravone was developed but failed in 2011 due to poor bioavailability. More recently, a sublingual (SL) tablet formulation of edaravone 30mg was developed by a Good Manufacturing Practices-compliant manufacturer in China. This study explored the bioavailability of the SL tablet of edaravone and aimed to provide evidence to support decision making in future clinical development. METHODS This 2-way crossover study was conducted in 10 healthy male volunteers. Eligible subjects were randomized, in a 1:1 ratio, to 1 of 2 dosing sequences: (1) SL edaravone 30mg, followed by edaravone 30mg IV infusion given over 30 minutes; or (2) edaravone 30mg IV infusion given over 30 minutes, followed by SL edaravone 30mg. The washout period between the 2 dosing periods was at least 24hours. Serial blood samples were collected in each dosing period. The bioavailability of the SL tablet was assessed using bioavailability analysis. Tolerability was evaluated throughout the study. FINDINGS The plasma concentration-time profile of the SL tablet was similar to that with the IV infusion. Amean (SD) Cmax of 2030.2 (517.2) ng/mL was reached within a median Tmax of 0.875hour, which was statistically significantly longer than the median Tmax with IV administration (0.5 hour). The Cmax with SL administration corresponded to 83.92% (90% CI, 73.22%-96.18%) of the Cmax with the start of IV infusion (2354.0 [336.6] ng/mL). The mean AUC0-t with SL dosing was 5420.07 (1429.75) h · ng/mL, which corresponded to 91.94% (90% CI, 86.81%-97.39%) of the AUC0-t with IV administration (5824.42 [1338.48] h · ng/mL). Two cases of adverse events were reported during the study; both were considered by the investigator to have been possibly not related to the study treatment. IMPLICATIONS The bioavailability of the SL tablet of edaravone was 91.94%. Compared with IV administration, Cmax with SL administration was ∼17% lower and Tmax was statistically significantly longer. The exposure differences can be addressed by modifying the strength of the SL tablet, and then conducting a second study to demonstrate the pharmacokinetic bioavailability of the sublingually administered new strength versus IV infusion of edaravone.
Collapse
Affiliation(s)
- Jiaqing Wang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People's Republic of China
| | - Xia Chen
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People's Republic of China.
| | - Baoshi Yuan
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People's Republic of China
| | - Weicong Wang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People's Republic of China
| | - Chunmin Xu
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People's Republic of China
| | - Weiwei Zhao
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People's Republic of China
| | - Pengpeng Zhao
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People's Republic of China
| | - Yilong Wang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People's Republic of China
| | - Xingquan Zhao
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People's Republic of China
| | - Yongjun Wang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China; Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
26
|
Xu W, Wen M, Yu J, Zhang Q, Polyakov NE, Dushkin AV, Su W. Mechanochemical preparation of kaempferol intermolecular complexes for enhancing the solubility and bioavailability. Drug Dev Ind Pharm 2018; 44:1924-1932. [PMID: 30035618 DOI: 10.1080/03639045.2018.1503292] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In this study, complexes of kaempferol (KF) with polysaccharide arabinogalactan (AG) and disodium glycyrrhizinate (Na2GA) were prepared through mechanochemical technique to improve the solubility and bioavailability of KF. The physicochemical properties and the interactions of KF with AG/Na2GA were investigated through dissolution, SEM, XRD, and DSC studies. The reduction of particle sizes and destruction of crystal forms revealed the formation of solid dispersion which may have assisted the dissolution of the drug. The accelerated stability study showed higher stability for KF-Na2GA complex. In vivo pharmacokinetic study was performed to observe the plasma drug concentrations for KF complexes. Mechanochemical complexation of KF with AG/Na2GA improved the pharmacological activity as evident by the inhibitory potential of the complexes towards carbohydrate metabolic enzymes. In vivo studies were performed in STZ-induced diabetic mice, where the group treated with KF-AG complex showed better liver and kidney function and lower blood glucose levels than pure KF. Therefore, mechanochemical complexes of KF with polysaccharide or glycyrrhizate may serve as a promising formulation for the treatment of diabetes.
Collapse
Affiliation(s)
- Wenhao Xu
- a National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals , Zhejiang University of Technology , Hangzhou , China
| | - Murong Wen
- a National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals , Zhejiang University of Technology , Hangzhou , China
| | - Jingbo Yu
- a National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals , Zhejiang University of Technology , Hangzhou , China
| | - Qihong Zhang
- a National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals , Zhejiang University of Technology , Hangzhou , China
| | - Nikolay E Polyakov
- b Institute of Chemical Kinetics and Combustion of Siberian Branch of Russian Academy of Sciences , Novosibirsk , Russia
| | - Alexandr V Dushkin
- a National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals , Zhejiang University of Technology , Hangzhou , China.,c Institute of Solid State Chemistry and Mechanochemistry , Novosibirsk , Russia
| | - Weike Su
- a National Engineering Research Center for Process Development of Active Pharmaceutical Ingredients, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals , Zhejiang University of Technology , Hangzhou , China.,d Key Laboratory for Green Pharmaceutical Technologies and Related Equipment of Ministry of Education, College of Pharmaceutical Sciences , Zhejiang University of Technology , Hangzhou , PR China
| |
Collapse
|
27
|
Physicochemical characteristics of the complexes of simvastatin and atorvastatin calcium with hydroxypropyl- β -cyclodextrin produced by mechanochemical activation. J Drug Deliv Sci Technol 2018. [DOI: 10.1016/j.jddst.2018.05.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
28
|
Parikh A, Kathawala K, Li J, Chen C, Shan Z, Cao X, Wang YJ, Garg S, Zhou XF. Self-nanomicellizing solid dispersion of edaravone: part II: in vivo assessment of efficacy against behavior deficits and safety in Alzheimer's disease model. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:2111-2128. [PMID: 30022810 PMCID: PMC6042531 DOI: 10.2147/dddt.s161944] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Alzheimer’s disease (AD) is a devastating neurodegenerative disorder that lacks any disease-modifying drug for the prevention and treatment. Edaravone (EDR), an approved free radical scavenger, has proven to have potential against AD by targeting multiple key pathologies including amyloid-beta (Aβ), tau phosphorylation, oxidative stress, and neuroinflammation. To enable its oral use, novel edaravone formulation (NEF) was previously developed. The aim of the present investigation was to evaluate safety and efficacy of NEF by using in vitro/in vivo disease model. Materials and methods In vitro therapeutic potential of NEF over EDR was studied against the cytotoxicity induced by copper metal ion, H2O2 and Aβ42 oligomer, and cellular uptake on SH-SY5Y695 amyloid-β precursor protein (APP) human neuroblastoma cell line. For in vivo safety and efficacy assessment, totally seven groups of APP/PS1 (five treatment groups, one each as a basal and sham control) and one group of C57BL/6 mice as a positive control for behavior tests were used. Three groups were orally treated for 3 months with NEF at an equivalent dose of EDR 46, 138, and 414 µmol/kg, whereas one group was supplied with each Donepezil (5.27 µM/kg) and Soluplus (amount present in NEF of 414 µmol/kg dose of EDR). Behavior tests were conducted to assess motor function (open-field), anxiety-related behavior (open-field), and cognitive function (novel objective recognition test, Y-maze, and Morris water maze). For the safety assessment, general behavior, adverse effects, and mortality were recorded during the treatment period. Moreover, biochemical, hematological, and morphological parameters were determined. Results Compared to EDR, NEF showed superior cellular uptake and neuroprotective effect in SH-SY5Y695 APP cell line. Furthermore, it showed nontoxicity of NEF up to 414 µM/kg dose of EDR and its potential to reverse AD-like behavior deficits of APP/PS1 mice in a dose-dependent manner. Conclusion Our results indicate that oral delivery of NEF holds a promise as a safe and effective therapeutic agent for AD.
Collapse
Affiliation(s)
- Ankit Parikh
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| | - Krishna Kathawala
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| | - Jintao Li
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ; .,Neuroscience Institute, Kunming Medical University, Kunming, Yunnan, China
| | - Chi Chen
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ; .,Central Laboratory, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhengnan Shan
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| | - Xia Cao
- Central Laboratory, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yan-Jiang Wang
- Department of Neurology, Daping Hospital, Third Military Medical University, Chongqing, Sichuan, China
| | - Sanjay Garg
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| |
Collapse
|
29
|
Parikh A, Kathawala K, Tan CC, Garg S, Zhou XF. Self-nanomicellizing solid dispersion of edaravone: part I - oral bioavailability improvement. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:2051-2069. [PMID: 30013324 PMCID: PMC6038876 DOI: 10.2147/dddt.s161940] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Edaravone (EDR) is known for its free radical scavenging, antiapoptotic, antinecrotic, and anticytokine effects in neurological and non-neurological diseases. It is currently available clinically as Radicava® and Radicut®, intravenous medications, recently approved for the treatment of amyotrophic lateral sclerosis and cerebral infarction. However, the oral use of EDR is still restricted by its poor oral bioavailability (BA) due to poor aqueous solubility, stability, rapid metabolism, and low permeability. The present study reports the development of novel EDR formulation (NEF) using self-nanomicellizing solid dispersion (SNMSD) strategy with the aim to enable its oral use. Materials and methods The selection of a suitable carrier for the development of NEF was performed based on the miscibility study. The optimization of EDR-to-carrier ratio was conducted via kinetic solubility study after preparing SNMSDs using solvent evaporation technique. The drug–polymer carrier interaction and self-nanomicellizing properties of NEF were investigated with advanced characterization studies. In vitro permeation, metabolism, and dissolution study was carried out to examine the effect of the presence of a carrier on physico-chemical properties of EDR. Additionally, the dose-dependent pharmacokinetic study of NEF was conducted and compared with the EDR suspension. Results Soluplus® (SOL) as a carrier was selected based on the potential for improving aqueous solubility. The NEF containing EDR and SOL (1:5) resulted in the highest enhancement in aqueous solubility (17.53-fold) due to amorphization, hydrogen bonding interaction, and micellization. Moreover, the NEF demonstrated significant improvement in metabolism, permeability, and dissolution profile of EDR. Furthermore, the oral BA of NEF showed 10.2-, 16.1-, and 14.8-fold enhancement compared to EDR suspension at 46, 138, and 414 µmol/kg doses. Conclusion The results demonstrated that SNMSD strategy could serve as a promising way to enhance EDR oral BA and NEF could be a potential candidate for the treatment of diseases in which oxidative stress plays a key role in their pathogenesis.
Collapse
Affiliation(s)
- Ankit Parikh
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| | - Krishna Kathawala
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| | - Chun Chuan Tan
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| | - Sanjay Garg
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia, ;
| |
Collapse
|
30
|
Li Q, Huang W, Yang J, Wang J, Hu M, Mo J, Cheng Y, Ou Z, Zhang ZJ, Guan S. Gastric retention pellets of edaravone with enhanced oral bioavailability: Absorption mechanism, development, and in vitro/in vivo evaluation. Eur J Pharm Sci 2018; 119:62-69. [PMID: 29630939 DOI: 10.1016/j.ejps.2018.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 03/11/2018] [Accepted: 04/01/2018] [Indexed: 12/12/2022]
Abstract
Absorption mechanism of edaravone (EDR) was studied to inform the preparation of gastric retention pellets with the aim to enhance its oral bioavailability. Three different models, namely, Caco-2 cells model, in situ single-pass intestinal perfusion model, and everted gut sac model in rats, were employed to characterize the gastrointestinal absorption kinetics of EDR. And it was found that passive transfer plays a vital role for the transport of EDR, and acidic condition is preferable for EDR absorption. Further, it is likely that EDR acts as a substrate for P-glycoprotein and multidrug-resistance protein. And hence, an orally available gastric retention pellets were developed accordingly. Pharmacokinetic experiments performed with rats and beagles showed that the absolute bioavailability of EDR solution and enteric-coated pellets following oral administration were 33.85% ± 2.45% and 7.64% ± 1.03%, indicating that stomach absorption is better than intestinal adsorption for EDR. However, the gastric retention pellets resulted in 68.96% absolute bioavailability and about 200% relative bioavailability in comparison to EDR solution, which was 9 times that of enteric-coated pellets. The present work demonstrates that gastric retention pellets has excellent potential as oral administration route for EDR.
Collapse
Affiliation(s)
- Qingguo Li
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Wenhai Huang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Juan Yang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Jianfeng Wang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Min Hu
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Jianmei Mo
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Yuzhu Cheng
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Zhanlun Ou
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Zhenyu Jason Zhang
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Shixia Guan
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| |
Collapse
|
31
|
Kong R, Zhu X, Meteleva ES, Chistyachenko YS, Suntsova LP, Polyakov NE, Khvostov MV, Baev DS, Tolstikova TG, Yu J, Dushkin AV, Su W. Enhanced solubility and bioavailability of simvastatin by mechanochemically obtained complexes. Int J Pharm 2017; 534:108-118. [DOI: 10.1016/j.ijpharm.2017.10.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 09/19/2017] [Accepted: 10/06/2017] [Indexed: 10/18/2022]
|
32
|
Porat D, Dahan A. Active intestinal drug absorption and the solubility-permeability interplay. Int J Pharm 2017; 537:84-93. [PMID: 29102702 DOI: 10.1016/j.ijpharm.2017.10.058] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 01/08/2023]
Abstract
The solubility-permeability interplay deals with the question: what is the concomitant effect on the drug's apparent permeability when increasing the apparent solubility with a solubility-enabling formulation? The solubility and the permeability are closely related, exhibit certain interplay between them, and ongoing research throughout the past decade shows that treating the one irrespectively of the other may be insufficient. The aim of this article is to provide an overview of the current knowledge on the solubility-permeability interplay when using solubility-enabling formulations for oral lipophilic drugs, highlighting active permeability aspects. A solubility-enabling formulation may affect the permeability in opposite directions; the passive permeability may decrease as a result of the apparent solubility increase, according to the solubility-permeability tradeoff, but at the same time, certain components of the formulation may inhibit/saturate efflux transporters (when relevant), resulting in significant apparent permeability increase. In these cases, excipients with both solubilizing and e.g. P-gp inhibitory properties may lead to concomitant increase of both the solubility and the permeability. Intelligent development of such formulation will account for the simultaneous effects of the excipients' nature/concentrations on the two arms composing the overall permeability: the passive and the active arms. Overall, thorough mechanistic understanding of the various factors involved in the solubility-permeability interplay may allow developing better solubility-enabling formulations, thereby exploiting the advantages analyzed in this article, offering oral delivery solution even for BCS class IV drugs.
Collapse
Affiliation(s)
- Daniel Porat
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Arik Dahan
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| |
Collapse
|
33
|
Parikh A, Kathawala K, Tan CC, Garg S, Zhou XF. Lipid-based nanosystem of edaravone: development, optimization, characterization and in vitro/in vivo evaluation. Drug Deliv 2017; 24:962-978. [PMID: 28633547 PMCID: PMC8241028 DOI: 10.1080/10717544.2017.1337825] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/23/2017] [Accepted: 05/30/2017] [Indexed: 12/25/2022] Open
Abstract
Edaravone (EDR) is a well-recognized lipophilic free radical scavenger for diseases including neurodegenerative disease, cardiovascular disease, and cancer. However, its oral use is restricted due to poor oral bioavailability (BA). The aim of present research was to enable its oral use by developing a lipid-based nanosystem (LNS). The components of LNS including oil, surfactants, and co-surfactants were selected based on their potential to maximize the solubilization in gastrointestinal (GI) fluids, reduce its glucuronidation and improve transmembrane permeability. The liquid LNS (L-LNS) with Capryol™ PGMC (Oil), Cremophor® RH 40:Labrasol®:TPGS 1000 (1:0.8:0.2) (Surfactant) and Transcutol P® (Co-surfactant) were optimized to form microemulsion having droplet size (16.25 nm), polydispersity index (0.039), % Transmittance (99.85%), and self-emulsification time (32 s). It significantly improved the EDR loading as well as its metabolism and permeability profile during transport across the GI tract. To overcome the possible drawbacks of L-LNS, Aerosil® 200 was used to formulate solid LNS (S-LNS), and its concentration was optimized based on flow properties. S-LNS possessed all quality attributes of L-LNS confirmed by solid-state characterization, reconstitution ability, and stability study. The dissolution rate of EDR was significantly enhanced with L-LNS and S-LNS in simulated gastric, and intestinal fluids. The pharmacokinetic study revealed significant improvement in relative BA, Cmax, and t1/2 with L-LNS and S-LNS against EDR suspension. Moreover, S-LNS showed superior cellular uptake and neuroprotective effect compared to EDR in SH-SY5Y695 cell line. An appropriate selection of the components of LNS could enable effective oral delivery of challenging therapeutics that are conventionally used by the parenteral administration.
Collapse
Affiliation(s)
- Ankit Parikh
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Krishna Kathawala
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Chun Chuan Tan
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Sanjay Garg
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, Division of Health Sciences, University of South Australia, Adelaide, Australia
| | - Xin-Fu Zhou
- School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, Division of Health Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
34
|
Singh MS, Tammam SN, Shetab Boushehri MA, Lamprecht A. MDR in cancer: Addressing the underlying cellular alterations with the use of nanocarriers. Pharmacol Res 2017; 126:2-30. [PMID: 28760489 DOI: 10.1016/j.phrs.2017.07.023] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 06/29/2017] [Accepted: 07/26/2017] [Indexed: 01/02/2023]
Abstract
Multidrug resistance (MDR) is associated with a wide range of pathological changes at different cellular and intracellular levels. Nanoparticles (NPs) have been extensively exploited as the carriers of MDR reversing payloads to resistant tumor cells. However, when properly formulated in terms of chemical composition and physicochemical properties, NPs can serve as beyond delivery systems and help overcome MDR even without carrying a load of chemosensitizers or MDR reversing molecular cargos. Whether serving as drug carriers or beyond, a wise design of the nanoparticulate systems to overcome the cellular and intracellular alterations underlying the resistance is imperative. Within the current review, we will initially discuss the cellular changes occurring in resistant cells and how such changes lead to chemotherapy failure and cancer cell survival. We will then focus on different mechanisms through which nanosystems with appropriate chemical composition and physicochemical properties can serve as MDR reversing units at different cellular and intracellular levels according to the changes that underlie the resistance. Finally, we will conclude by discussing logical grounds for a wise and rational design of MDR reversing nanoparticulate systems to improve the cancer therapeutic approaches.
Collapse
Affiliation(s)
- Manu S Singh
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany
| | - Salma N Tammam
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany; Department of Pharmaceutical Technology, German University of Cairo, Egypt
| | | | - Alf Lamprecht
- Department of Pharmaceutical Technology and Biopharmceutics, University of Bonn, Germany; Laboratory of Pharmaceutical Engineering (EA4267), University of Franche-Comté, Besançon, France.
| |
Collapse
|
35
|
Eleuteri C, Olla S, Veroni C, Umeton R, Mechelli R, Romano S, Buscarinu MC, Ferrari F, Calò G, Ristori G, Salvetti M, Agresti C. A staged screening of registered drugs highlights remyelinating drug candidates for clinical trials. Sci Rep 2017; 7:45780. [PMID: 28387380 PMCID: PMC5384285 DOI: 10.1038/srep45780] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/01/2017] [Indexed: 12/13/2022] Open
Abstract
There is no treatment for the myelin loss in multiple sclerosis, ultimately resulting in the axonal degeneration that leads to the progressive phase of the disease. We established a multi-tiered platform for the sequential screening of drugs that could be repurposed as remyelinating agents. We screened a library of 2,000 compounds (mainly Food and Drug Administration (FDA)-approved compounds and natural products) for cellular metabolic activity on mouse oligodendrocyte precursors (OPC), identifying 42 molecules with significant stimulating effects. We then characterized the effects of these compounds on OPC proliferation and differentiation in mouse glial cultures, and on myelination and remyelination in organotypic cultures. Three molecules, edaravone, 5-methyl-7-methoxyisoflavone and lovastatin, gave positive results in all screening tiers. We validated the results by retesting independent stocks of the compounds, analyzing their purity, and performing dose-response curves. To identify the chemical features that may be modified to enhance the compounds' activity, we tested chemical analogs and identified, for edaravone, the functional groups that may be essential for its activity. Among the selected remyelinating candidates, edaravone appears to be of strong interest, also considering that this drug has been approved as a neuroprotective agent for acute ischemic stroke and amyotrophic lateral sclerosis in Japan.
Collapse
Affiliation(s)
- C. Eleuteri
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - S. Olla
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato 09042, Italy
| | - C. Veroni
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - R. Umeton
- Center for Experimental Neurological Therapies, Sant’Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Italy
| | - R. Mechelli
- Center for Experimental Neurological Therapies, Sant’Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Italy
| | - S. Romano
- Center for Experimental Neurological Therapies, Sant’Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Italy
| | - MC. Buscarinu
- Center for Experimental Neurological Therapies, Sant’Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Italy
| | - F. Ferrari
- Department of Medical Science, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, 44121 Ferrara, Italy
| | - G. Calò
- Department of Medical Science, Section of Pharmacology and National Institute of Neuroscience, University of Ferrara, 44121 Ferrara, Italy
| | - G. Ristori
- Center for Experimental Neurological Therapies, Sant’Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Italy
| | - M. Salvetti
- Center for Experimental Neurological Therapies, Sant’Andrea Hospital, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Italy
- IRCCS Istituto Neurologico Mediterraneo (INM) Neuromed, 86077 Pozzilli, IS, Italy
| | - C. Agresti
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy
| |
Collapse
|
36
|
Parikh A, Kathawala K, Tan CC, Garg S, Zhou XF. Development of a novel oral delivery system of edaravone for enhancing bioavailability. Int J Pharm 2016; 515:490-500. [DOI: 10.1016/j.ijpharm.2016.10.052] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 10/14/2016] [Accepted: 10/23/2016] [Indexed: 12/18/2022]
|
37
|
Aljhni R, Andre C, Lethier L, Guillaume YC. An HPLC chromatographic framework to analyze the β-cyclodextrin/solute complexation mechanism using a carbon nanotube stationary phase. Talanta 2015; 144:226-32. [DOI: 10.1016/j.talanta.2015.06.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 06/03/2015] [Accepted: 06/05/2015] [Indexed: 11/30/2022]
|
38
|
Vieira ACC, Ferreira Fontes DA, Chaves LL, Alves LDS, de Freitas Neto JL, de La Roca Soares MF, Soares-Sobrinho JL, Rolim LA, Rolim-Neto PJ. Multicomponent systems with cyclodextrins and hydrophilic polymers for the delivery of Efavirenz. Carbohydr Polym 2015; 130:133-40. [DOI: 10.1016/j.carbpol.2015.04.050] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 04/21/2015] [Accepted: 04/23/2015] [Indexed: 01/07/2023]
|
39
|
Zhang X, Chen G, Zhang T, Ma Z, Wu B. Effects of PEGylated lipid nanoparticles on the oral absorption of one BCS II drug: a mechanistic investigation. Int J Nanomedicine 2014; 9:5503-14. [PMID: 25473287 PMCID: PMC4251747 DOI: 10.2147/ijn.s73340] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Lipid nanocarriers are becoming a versatile platform for oral delivery of lipophilic drugs. In this article, we aimed to explore the gastrointestinal behaviors of lipid nanoparticles and the effect of PEGylation on oral absorption of fenofibrate (FN), a Biopharmaceutics Classification System (BCS) II model drug. FN-loaded PEGylated lipid nanoparticles (FN-PLNs) were prepared by the solvent-diffusion method and characterized by particle size distribution, morphology, Fourier transform infrared spectroscopy, and drug release. Lipolytic experiments were performed to assess the resistance of lipid nanoparticles against pancreatic lipase. Pharmacokinetics was evaluated in rats after oral administration of FN preparations. The obtained FN-PLNs were 186.7 nm in size with an entrapment efficiency of >95%. Compared to conventional lipid nanoparticles, PLNs exhibited slower drug release in the lipase-containing medium, strikingly reduced mucin binding, and suppressed lipolysis in vitro. Further, oral absorption of FN was significantly enhanced using PLNs with relative bioavailability of 123.9% and 157.0% to conventional lipid nanoparticles and a commercial formulation (Lipanthyl®), respectively. It was demonstrated that reduced mucin trapping, suppressed lipolysis, and/or improved mucosal permeability were responsible for increased oral absorption. These results facilitated a better understanding of the in vivo fate of lipid nanoparticles, and suggested the potential of PLNs as oral carriers of BCS II drugs.
Collapse
Affiliation(s)
- Xingwang Zhang
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People's Republic of China
| | - Guijiang Chen
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People's Republic of China
| | - Tianpeng Zhang
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People's Republic of China
| | - Zhiguo Ma
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People's Republic of China
| | - Baojian Wu
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, People's Republic of China
| |
Collapse
|